You are on page 1of 11

Articles

Efficacy of live oral rotavirus vaccines by duration of


follow-up: a meta-regression of randomised controlled trials
Andrew Clark*, Kevin van Zandvoort*, Stefan Flasche, Colin Sanderson, Julie Bines, Jacqueline Tate, Umesh Parashar, Mark Jit

Summary
Background The duration of protection offered by rotavirus vaccines varies across the world, and this variation is Lancet Infect Dis 2019;
important to understanding and predicting the effects of the vaccines. There is now a large body of evidence on the 19: 717–27
efficacy of live oral rotavirus vaccines in different settings, but these data have never been synthesised to obtain robust Published Online
June 6, 2019
estimates of efficacy by duration of follow-up. Our aim is to estimate the efficacy of live oral rotavirus vaccines at each
http://dx.doi.org/10.1016/
point during follow-up and by mortality stratum. S1473-3099(19)30126-4
See Comment page 673
Methods In our meta-regression study, we identified all randomised controlled trials of rotavirus vaccines published *Contributed equally
until April 4, 2018, using the results of a Cochrane systematic review, and cross checked these studies against those
London School of Hygiene and
identified by another systematic review. We excluded trials that were based on special populations, trials without an Tropical Medicine, London, UK
infant schedule, and trials without clear reporting of numbers of enrolled infants and events in different periods of (A Clark PhD,
follow-up. For all reported periods of follow-up, we extracted the mean duration of follow-up (time since administration K van Zandvoort MSc,
S Flasche PhD,
of the final dose of rotavirus vaccination), the number of enrolled infants, and case counts for rotavirus-positive Prof C Sanderson PhD,
severe gastroenteritis in both non-vaccinated and vaccinated groups. We used a Bayesian hierarchical Poisson meta- Prof M Jit PhD); Murdoch
regression model to estimate the pooled cumulative vaccine efficacy (VE) and its waning with time for three mortality Children’s Research Institute,
strata. We then converted these VE estimates into instantaneous VE (iVE). Melbourne, VIC, Australia
(Prof J Bines MD); Department
of Paediatrics, The University
Findings In settings with low mortality (15 observations), iVE pooled for infant schedules of Rotarix and RotaTeq was of Melbourne, Melbourne, VIC,
98% (95% credibility interval 93–100) 2 weeks following the final dose of vaccination and 94% (87–98) after 12 months. Australia (Prof J Bines);
In medium-mortality settings (11 observations), equivalent estimates were 82% (74–92) after 2 weeks and 77% (67–84) Department of
Gastroenterology and Clinical
after 12 months. In settings with high mortality (24 observations), there were five different vaccines with observation Nutrition, Royal Children’s
points for infant schedules. The pooled iVE was 66% (48–81) after 2 weeks of follow-up and 44% (27–59) after Hospital, Melbourne, VIC,
12 months. Australia (Prof J Bines); Centers
for Disease Control and
Prevention, Atlanta, GA, USA
Interpretation Rotavirus vaccine efficacy is lower and wanes more rapidly in high-mortality settings than in (J Tate PhD, U Parashar MD); and
low-mortality settings, but the earlier peak age of disease in high-mortality settings means that live oral rotavirus Modelling and Economics Unit,
vaccines are still likely to provide substantial benefit. Public Health England, London,
UK (Prof M Jit)

Funding Bill & Melinda Gates Foundation. Correspondence to:


Dr Andrew Clark, London School
of Hygiene and Tropical
Copyright © 2019 The Author(s). Published by Elsevier Ltd. This is an Open Access article under the CC BY 4.0 license. Medicine, London WC1E 7HT, UK
andrew.clark@lshtm.ac.uk
Introduction of India, Pune, India), and RV3-BB (Murdoch Children’s
Rotavirus gastroenteritis is estimated to cause around Research Institute, Melbourne, Australia), but these
200 000 child deaths each year,1–3 mostly in sub-Saharan vaccines have also reported low or waning efficacy in high-
Africa and south Asia. Episodes of rotavirus gastroenteritis mortality settings (eg, India, Indonesia, and Niger) when
occur frequently in young children irrespective of living used as part of a standard infant schedule.13–16 Alternative
standards and are a major contributor to health-care costs schedules are being considered as one way to improve
worldwide.4,5 efficacy in the second year of life. Alternatives might
More than half of the countries in the world have involve administering the first dose at birth15 or
introduced rotavirus vaccines into their national administering a booster dose at age 9–12 months.17
immunisation programmes.6 Infants typically receive two Countries considering the introduction of rotavirus
oral doses of Rotarix (GlaxoSmithKline Biologicals, vaccine, global bodies such as WHO, and donors funding
London, UK) or three oral doses of RotaTeq (Merck & Co, vaccine introduction in resource-poor settings require
Kenilworth, NJ, USA) in the first 6 months of life.7,8 Both accurate projections of the potential effect of vaccination.
vaccines have shown high and durable efficacy against Such projections are also useful in surveillance after
episodes of severe rotavirus gastroenteritis in high-income vaccine introduction, to ensure that the vaccine is
settings but lower and less durable efficacy in sub-Saharan performing as expected and to estimate the remaining
Africa and south Asia.9–12 Other live oral rotavirus vaccines burden of disease after the vaccine has been introduced.
are becoming available, such as ROTAVAC (Bharat Mathematical models can predict the potential effect of
Biotech, Hyderabad, India), ROTASIIL (Serum Institute rotavirus vaccines but require credible estimates of vaccine

www.thelancet.com/infection Vol 19 July 2019 717


Articles

Research in context
Evidence before the study efficacy is lower and wanes more rapidly in high-mortality
In a Cochrane systematic review published in 2012, settings than in low-mortality settings. We show that, in
Soares-Weiser and colleagues identified 11 randomised Indonesia, a neonatal schedule provides more durable
controlled trials that showed that live oral rotavirus vaccines protection than the standard infant schedule, although this
induce high and durable efficacy against episodes of severe analysis was based on very few case counts in each week of
rotavirus gastroenteritis in high-income settings, but lower follow-up.
and less durable efficacy in sub-Saharan Africa and south Asia.
Implications of all the available evidence
Added value of this study Live oral rotavirus vaccines are likely to provide substantial
To our knowledge, this is the first study in which all the benefit globally. In high-mortality settings, strategies to
available evidence from randomised controlled trials has been optimise the effect of rotavirus vaccination warrant serious
synthesised to obtain robust estimates of efficacy by duration consideration. Estimates of the instantaneous efficacy of live
of follow-up. We include several new data points from Asia oral rotavirus vaccines by duration of follow-up will be crucial
and use a novel approach to convert cumulative vaccine efficacy to understanding the potential effect of alternative rotavirus
into instantaneous vaccine efficacy. Our analysis provides the vaccination schedules in different countries.
most comprehensive evidence to date that rotavirus vaccine

efficacy by duration of follow-up in different settings. This periods of follow-up. The outcome measure was efficacy
information is also crucial to the evaluation of alternative against episodes of severe rotavirus gastroenteritis, which
vaccination schedules. A large body of evidence now exists is the primary endpoint reported in nearly all RCTs of
from high-quality randomised controlled trials (RCTs) in rotavirus vaccines. Severe rotavirus gastroenteritis is
different parts of the world, but these data have never been defined as 11–20 points on the Vesikari scale,20 or for
pooled and synthesised to obtain robust estimates of some older trials, 15–24 points on the Clark scale.21 If this
vaccine efficacy by duration of follow-up. Combining this outcome was not reported, we used the closest available
evidence is not straightforward. There is substantial proxy, such as efficacy against episodes of rotavirus
variation in trial settings, follow-up periods, sample sizes, gastroenteritis, which involved admission to hospital or
case definitions, and statistical methods used to calculate the emergency department. In all studies, rotavirus-
CIs. In addition, the main outcome reported in RCTs is the positive episodes were detected by enzyme immunoassay.
cumulative vaccine efficacy (VE) over a period of many This study was approved by the ethics committee
weeks, but if there is evidence of vaccine waning, then (Ref 15829) of the London School of Hygiene and Tropical
the cumulative efficacy over the entire follow-up period Medicine.
might be different to the actual instantaneous VE (iVE) at
different times within that period of follow-up. Follow-up definition
Our aim is to estimate the instantaneous efficacy of We extracted vaccine efficacy for all reported periods of
live oral rotavirus vaccines by duration of follow-up (time follow-up. Most of the studies reported results at two
since administration of the final dose of rotavirus follow-up points. However, we also included studies with
vaccination) and by mortality strata. a single follow-up point. We extracted the number of
individuals eligible for per-protocol analysis and the
Methods number of rotavirus-positive cases in both the non-
Search strategy and selection criteria vaccinated and vaccinated groups. We also extracted the
We ran a meta-regression, in which we included all mean duration of follow-up in months. We extracted per-
individual RCTs that were identified in a Cochrane protocol estimates because they exclude any disease cases
systematic review of studies published until April 8, 2018,18 reported in the first 14 days after vaccination and include
and cross-checked the list against the studies identified by only infants who received all recommended doses. We
a review by Lamberti and colleagues.19 AC obtained the added 14 days to the reported mean duration of follow-up
list of RCTs from the authors of the Cochrane review, to calculate the entire period between administration of
cross-checked the list against the studies identified by a the last dose and the mean age at follow-up. If all infants
review by Lamberti and colleagues, extracted relevant in a study were followed up to a specific age (eg, 12 months),
data, and contacted the lead investigators of the study we subtracted the mean age of administration of the final
where further clarification was needed. A full assessment dose (or target age if the mean was not reported) from the
of the risks of bias associated with each rotavirus vaccine specific follow-up age.
efficacy trial is described in detail in the Cochrane review.18
We excluded trials that were based on special populations, Stratification of studies
trials without an infant schedule, and trials without clear To account for heterogeneity between the RCT sites, we
reporting of enrolled infants and events in different grouped all 201 countries in the world into quintiles

718 www.thelancet.com/infection Vol 19 July 2019


Articles

(very low mortality, low mortality, medium mortality, t months of follow-up. We used the log of the person-
high mortality, and very high mortality) using the time to estimate the log of the number of cases, so that
under-5 mortality rate reported for the period 2010–15 in the person-time was used on its identity scale when
the 2017 revision of the UN Population Division database.22 converting the cases to the identity scale. Total person-
We further collapsed the very low and low quintiles and months of follow-up was calculated as the number of
the high and very high quintiles to give three strata participants at the beginning of the follow-up period
for deaths under age 5 years per 1000 livebirths: multiplied by the reported mean duration of follow-up.
low (<13·5 deaths per 1000 livebirths), medium The hierarchical component of the model ensured that
(13·5–28·1 deaths per 1000 livebirths), and high parameter values of the study-specific RR were identical
(>28·1 deaths per 1000 livebirths). Each RCT was then across periods in studies with more than one data point
assigned to a specific stratum. For RCTs with multiple (eg, RR for period one and RR for period one plus two
sites across several countries, we included each individual combined).
country as a separate observation point when this was We estimated best-fitting model parameters using
possible. If RCT results were not disaggregated by country, Markov Chain Monte Carlo methods. Gibbs sampling was
we used the sample size in each site to calculate a weighted used to draw from posterior distributions. We used non-
under-5 mortality rate and used this estimate to assign the informative prior distributions for all parameters. We
trial to a specific mortality stratum. We restricted the ran four parallel Markov Chain Monte Carlo chains and
pooled analysis to infant schedules only. visually assessed whether chains converged. We report
medians and 95% credible intervals from the posterior
Recalculating cumulative vaccine efficacy for reported distributions of the cumulative VE. In the absence of any
periods of follow-up prior knowledge about the probable shape of waning, we
We observed substantial variation in the way authors explored several functional forms, including linear, power
estimated VE and 95% confidence intervals. Our pooled law, sigmoid, and gamma (appendix p 8). We assessed See Online for appendix
analyses used case counts and sample sizes reported in their goodness of fit using the deviance information
trials to generate credible intervals, but to ensure criterion (DIC), visual assessment, and biological
consistent reporting of the data in the summary table plausibility.
and plots, we also recalculated VE and 95% confidence The best-fitting function of VE by duration of follow-up
intervals using the method of Daly and Altman.23,24 was used to estimate the iVE by duration of follow-up
VE was calculated as 1  – 
relative risk (RR) with using a novel approach based on Kaplan-Meier survival
zero inflation to 0·5.25 estimates. More details about the method, including a
derivation of the method, are provided in the appendix
Efficacy by duration of follow-up and mortality strata (p 1). The iVE at time t, termed 1 – σ(t), is retrieved using
We used a Bayesian hierarchical meta-regression model the following formula:
to estimate cumulative VE by duration of follow-up. We
generated separate pooled estimates for RCTs in low t–1 λ(x)
mortality, medium mortality, and high mortality strata. 1 – σ(t) = 1 – ϑ(t) + ∫ x = 0 (ϑ(t) – σ(x)) dx
λ(t)
We assumed that errors around the observed numbers of
cases in the unvaccinated and vaccinated groups followed
Poisson distributions. The total number of cases in the In this formula, iVE as a function of VE at time t is
unvaccinated group in study i and period p, termed Yi,p,u, termed 1 – σ(t), all iVEs up until time t are termed 1 – σ(x),
was estimated using the following generalised linear the baseline rate or force of infection at time t is termed
model: λ(t), and all baseline rates up until time t are termed λ(x).
ϑ denotes the relative rate, but we can only estimate
log(Yi,p,u) = λi,p + log (Pi,p,u) relative risks (θ) with our dataset. Severe rotavirus
gastroenteritis is a rare outcome, so we assume that θ is
approximately equal to ϑ.
Similarly, the total number of cases in the vaccinated iVE and VE are identical at time t=0, that is to say
group in study i and period p, termed Yi,p,v, was estimated 1 – σ(0) = 1 – ϑ(0). The formula can then be used to iterate
using: over all VEs to retrieve iVEs. If changes in baseline rates
are not known, they can be assumed to be similar over
log(Yi,p,v) = λi,p + log (Pi,p,v) + θi(ti,p) time, so that:

λ(x)
=1
in which λi,p is the baseline rate of becoming infected, λ(t)
Pi,p,v and Pi,p,u are the total person-months of follow-up in
the vaccinated and unvaccinated group, respectively, and For each stratum, we reported iVE at standard follow-
θi(ti,p) is the cumulative relative risk (RR) in study i, at up times. We calculated empirical p values and credible

www.thelancet.com/infection Vol 19 July 2019 719


Articles

intervals to investigate differences between strata. We and credible intervals to investigate differences between
ran a sensitivity analysis to calculate iVE with and without schedules.
observations from trials with large sample sizes.
Analyses were done using R version 3.4.326 and the Role of the funding source
rjags package.27 Code for the model and conversion The funder of the study had no role in study design, data
method is provided online.28 collection, data analysis, data interpretation, or writing
of this report. The corresponding author had full access
Head-to-head comparison of efficacy for alternative to all the data in the study and had final responsibility
schedules for the decision to submit for publication.
To compare the efficacy and waning associated with
different rotavirus vaccine schedules, we identified RCTs Results
that directly compared different vaccine schedules head We included 50 observation points from 50 observations
to head and requested more detailed unpublished published before April 4, 2018, in populations with low
information from the investigators on the number of under-5 mortality (15 observations), medium under-5
case counts and individuals occurring in each week of mortality (11 observations), and high under-5 mortality
follow-up after the last dose was administered. In sites (24 observations) (table 1). We excluded trials that
with available data, we fitted the same models as in the evaluated special groups, such as populations with high
pooled analysis but without the hierarchical parameters. HIV prevalence46 and breastfed infants.47 We excluded
These models used the same waning functions as in the the Finnish Extension Trial48,49 because the results could
pooled analysis (appendix, p 6) but with refitted not be disentangled from a pooled estimate for five
parameters. Again, VE by duration of follow-up was European countries reported separately.42 For the
converted to iVE, and we calculated empirical p values pooled analysis we focused on infant schedules, so

Schedule Vaccine brand Score* Mean follow-up Non-vaccinated group† Vaccinated group Cumulative efficacy
(months since (95% confidence
final dose) intervals)
Mean age Doses Cases Number of Cases Number of
(weeks) individuals individuals
at dose 1
High-mortality countries
Bangladesh9 10·0 2 Rotarix V11–20 8·1 35 301 9 292 73% (45 to 87)
Malawi10 11·0 2 Rotarix V11–20 7·7 38 483 21 525 49% (15 to 70)
Malawi10 11·0 2 Rotarix V11–20 15·6 53 483 38 525 34% (2 to 56)
South Africa‡11 11·0 2 Rotarix V11–20 7·7 9 408 5 418 46% (–60 to 82)
South Africa‡11 11·0 2 Rotarix V11–20 15·6 13 408 9 418 32% (–56 to 71)
Malawi10 6·2 3 Rotarix V11–20 7·7 38 483 20 505 50% (15 to 70)
Malawi10 6·2 3 Rotarix V11–20 15·6 53 483 32 505 42% (12 to 62)
South Africa‡11 6·2 3 Rotarix V11–20 7·7 9 408 1 425 89% (16 to 99)
South Africa‡11 6·2 3 Rotarix V11–20 15·6 13 408 2 425 85% (35 to 97)
Bangladesh12 8·3 3 RotaTeq V11–20 8·0 31 565 17 563 45% (2 to 69)
Bangladesh12 8·3 3 RotaTeq V11–20 14·7 56 565 33 563 41% (11 to 61)
Ghana29 8·4 3 RotaTeq V11–20 8·0 42 1081 15 1081 64% (36 to 80)
Ghana29 8·4 3 RotaTeq V11–20 14·5 57 1081 26 1081 54% (28 to 71)
Kenya29 7·3 3 RotaTeq V11–20 8·1 12 611 2 610 83% (26 to 96)
Kenya29 7·3 3 RotaTeq V11–20 12·3 14 611 5 610 64% (1 to 87)
Mali§29 6·9 3 RotaTeq V11–20 8·5 4 921 4 921 0% (–299 to 75)
Mali29 6·9 3 RotaTeq V11–20 14·9 58 921 48 921 17% (–20 to 43)
Niger14 6·8 3 ROTASIIL V11–20 5·6 87 1728 31 1780 65% (48 to 77)
India¶13 6·9 3 ROTASIIL V11–20 8·3 94 3498 61 3527 36% (11 to 53)
India13 6·9 3 ROTASIIL V11–20 20·0 275 3502 171 3533 38% (26 o 49)
India16 6·8 3 ROTAVAC V11–20 8·2 64 2187 56 4354 56% (37 to 69)
India16 6·8 3 ROTAVAC V11–20 13·4 76 2187 71 4354 53% (35 to 66)
Indonesia||15 9·3 3 RV3-BB V11–20 7·5 17 504 4 511 77% (32 to 92)
Indonesia||15 9·3 3 RV3-BB V11–20 13·5 28 504 14 511 51% (7 to 74)
(Table 1 continues on next page)

720 www.thelancet.com/infection Vol 19 July 2019


Articles

Schedule Vaccine brand Score* Mean follow-up Non-vaccinated group† Vaccinated group Cumulative efficacy
(months since (95% confidence
final dose) intervals)
Mean age Doses Cases Number of Cases Number of
(weeks) individuals individuals
at dose 1
(Continued from previous page)
Medium-mortality countries
China30 9·6 2 Rotarix V11–20 4·0 32 1573 8 1575 75% (46 to 88)
China30 9·6 2 Rotarix V11–20 16·5 75 1573 21 1575 72% (55 to 83)
Latin America31 (n=3) 8·4 2 Rotarix V11–20 7·5 34 454 27 1392 74% (58 to 84)
Latin America32 (n=6) 8·6 2 Rotarix V11–20 7·9 19 2099 7 4211 82% (56 to 92)
Latin America33 (n=10) 8·0 2 Rotarix V11–20 8·8 58 7081 10 7205 83% (67 to 91)
Latin America33 (n=10) 8·0 2 Rotarix V11–20 20·5 161 7081 32 7205 80% (71 to 87)
China34 8·5 3 RotaTeq V11–20 9·8 52 1946 11 1930 79% (59 to 89)
Latin America35 (n=5) 9·7 3 RotaTeq Hosp/ED 19·0 10 2237 1 2252 90% (22 to 99)
USA36 (Navajo) >6 3 RotaTeq C11–24 8·8 37 403 4 392 89% (69 to 96)
Vietnam12 9·7 3 RotaTeq V11–20 8·0 7 442 2 446 72% (–36 to 94)
Vietnam12 9·7 3 RotaTeq V11–20 12·3 15 442 5 446 67% (10 to 88)
Low-mortality countries
Europe37 (n=6) 11·5 2 Rotarix V11–20 5·3 60 1302 5 2572 96% (90 to 98)
Europe37 (n=6) 11·5 2 Rotarix V11–20 17·3 127 1302 24 2572 90% (85 to 94)
Finland38 8·3 2 Rotarix V11–20 5·3 5 123 1 245 90% (15 to 99)
Finland38 8·3 2 Rotarix V11–20 17·3 10 123 3 245 85% (46 to 96)
Japan39 7·7 2 Rotarix V11–20 20·6 12 250 2 498 92% (63 to 98)
Southeast Asia40 (n=3) 12·0 2 Rotarix V11–20 7·4 15 5256 0 5263 97% (46 to 100)
Southeast Asia40 (n=3) 12·0 2 Rotarix V11–20 31·7 64 5256 2 5263 97% (87 to 99)
Southeast Asia40 (n=3) 12·0 2 Rotarix V11–20 19·5 51 5256 2 5263 96% (84 to 99)
USA41 13·0 2 Rotarix All RVGE 7·0 18 107 2 108 89% (54 to 97)
Europe42 (n=5) 10·0 3 RotaTeq C17–24 13·3 43 1188 0 1120 99% (80 to 100)
Europe42 (n=5)¶ 10·0 3 RotaTeq C17–24 19·0 61 1155 1 1088 98% (87 to 100)
USA35 9·7 3 RotaTeq Hosp/ED 19·0 58 12 179 3 12 284 95% (84 to 98)
Finland and USA43 10·0 3 RotaTeq C17–24 4·4 6 661 0 651 92% (–38 to 100)
Japan44 7·6 3 RotaTeq C17–24 6·7 10 381 0 380 95% (19 to 100)
USA45 >8 3 RotaTeq C17–24 5·5 8 183 0 187 94% (1 to 100)
The cumulative efficacy for reported periods of follow-up after two or three doses of live oral rotavirus vaccines are shown. *Scores denote the points on the Vesikari scale 11–20 (V11–20) or Clark scale 17–24
(C17–24). All RVGE denotes any severity of rotavirus-positive gastroenteritis. Hosp/ED denotes rotavirus-positive hospitalisation or emergency department visit.†All randomised controlled trials were placebo
controlled with the exception of the Rotarix trial in Bangladesh. ‡Data only extracted for the South African cohort that was followed for two successive seasons. §There were surveillance issues in the first year of
trial in Mali that have been postulated to contribute to the low efficacy in the first period, but we did not adjust for this. ¶N values were adjusted to be the same for both follow-up periods in the Bayesian
meta-regression. ||For the neonatal schedule, the cumulative efficacy was 94% (95% CI 55–99) after about 9 months of follow-up and 75% (43–89) after about 15 months.

Table 1: Observations from published randomised controlled trials included in the pooled analysis of infant schedules

excluded the neonatal RotaShield trial50 in Ghana and We estimated VE and iVE (median and 95% credible
the neonatal schedule group of the RV3-BB trial15 in intervals) by duration of follow-up (figure 1, table 2). In
Indonesia. The neonatal schedule group of the settings with low mortality (15 observations), iVE pooled for
RV3-BB trial15 was included in a separate head-to-head infant schedules of Rotarix and RotaTeq was 98%
comparison of the infant and neonatal schedule in (95% credibility interval 93–100) 2 weeks following the final
Indonesia. dose of vaccination and 94% (87–98) after 12 months.
Most of the data points (41 [82%] of 50) were reported Equivalent pooled estimates for medium-mortality settings
using a Vesikari score of 11–20. There were 24 data points (11 observations) were 82% (74–92) after 2 weeks and
for Rotarix, 19 for RotaTeq, two for RV-3BB, three for 77% (67–84) after 12 months. In settings with high mortality
ROTASIIL, and two for ROTAVAC. More data points (24 observations), there were five vaccines with observation
(30 [60%] of 50) were based on a three-dose schedule points for infant schedules. The pooled iVE was 66% (48–81)
than a two-dose schedule (20 [40%] of 50). The mean age after 2 weeks of follow-up and 44% (27–59) after 12 months.
of administration for the first dose ranged from We found good evidence that iVE was significantly lower
6 weeks to 13 weeks. in medium-mortality settings than in low-mortality

www.thelancet.com/infection Vol 19 July 2019 721


Articles

Cumulative
Low mortality Medium mortality High mortality
100

• ••• ••

••
••
• • •
•• •
• •
80
• •• • • •
••

Vaccine efficacy (%)

60
• • • •

•• •• •
40
• ••
• •
20 •
0 •
−20

Instantaneous
100

80
Vaccine efficacy (%)

60

40

20

−20
2 52 104 152 2 52 104 152 2 52 104 152
Time since final dose of vaccination (weeks) Time since final dose of vaccination (weeks) Time since final dose of vaccination (weeks)

Figure 1: Median and 95% credible intervals of cumulative and instantaneous vaccine efficacy by duration of follow-up and setting after two or three doses
of oral rotavirus vaccination (infant schedules only)
A simple power function was used to represent vaccine waning over time; equivalent plots based on other potential waning functions are available in the appendix
(p 10). Each blue dot represents the VE for each observation. The size of the dot represents the relative sample size of the study. The error bars represent 95% CIs
around the VE. VE=vaccine efficacy.

Low-mortality Medium-mortality High-mortality High-mortality India


countries countries countries countries (except India)
2 weeks 98% (93 to 100) 82% (74 to 92) 66% (48–81) 81% (56–94) 54% (–78 to 88)
1 month 98% (93 to 100) 81% (74 to 90) 62% (47–75) 74% (53–88) 52% (–89 to 88)
2 months 97% (93 to 99) 80% (73 to 87) 57% (45–67) 66% (50–79) 49% (–105 to 87)
3 months 96% (92 to 99) 79% (73 to 86) 54% (44–64) 61% (48–72) 48% (–108 to 86)
6 months 95% (91 to 98) 78% (71 to 84) 49% (40–61) 49% (38–64) 45% (–115 to 86)
9 months 95% (89 to 98) 77% (69 to 84) 46% (33–60) 42% (22–61) 43% (–124 to 86)
12 months 94% (87 to 98) 77% (67 to 84) 44% (27–59) 36% (5–60) 42% (–128 to 85)
18 months 94% (83 to 97) 77% (63 to 84) 41% (17–58) 27% (–26 to 59) 41% (–135 to 85)
24 months 93% (79 to 97) 76% (59 to 83) 38% (9–58) 19% (–54 to 57) 40% (–139 to 85)
36 months 92% (69 to 97) 76% (53 to 83) 35% (–4 to 57) 7% (–107 to 56) 39% (–149 to 85)
48 months 91% (58 to 97) 75% (48 to 83) 32% (–14 to 57) –2% (–154 to 56) 38% (–154 to 85)
60 months 91% (48 to 97) 75% (44 to 83) 30% (–23 to 57) –10% (–200 to 55) 37% (–163 to 85)

Table 2: Median instantaneous vaccine efficacy and 95% credible intervals by duration of follow-up and setting after two or three doses of oral rotavirus
vaccination (infant schedules only)

settings after 2 weeks and 12 months of follow-up. The stratum. Given their large sample sizes, we investigated
median absolute percentage point difference in iVE was whether these two studies were driving the results in the
16% (95% credibility interval 4–24, p=0·0023) at 2 weeks high-mortality setting. Therefore, we ran a sensitivity
and 17% (8–28, p=0·0022) at 12 months. We found weak analysis to calculate iVE with and without the Indian
evidence that iVE was significantly lower in high-mortality data points, and for India alone (table 2; appendix p 27).
settings compared with medium-mortality settings after We found no evidence that iVE significantly differed
2 weeks and strong evidence that iVE was lower after after 2 weeks or 12 months of follow-up when excluding
12 months. The median difference was 16% (95% credibility the Indian data points from the high-mortality stratum.
interval –1 to 38, p=0·089) at 2 weeks and 33% (15 to 51, The median difference absolute percentage point differ­
p=0·0011) at 12 months. ence in iVE was –13% (95% credibility interval –39 to 16,
Two large studies in India (of ROTAVAC16 and p=0·81) after 2 weeks and 8% (–22 to 42, p=0·31) after
ROTASIIL)13 were included in the high-mortality 12 months.

722 www.thelancet.com/infection Vol 19 July 2019


Articles

Cumulative Instantaneous
100 ••••••••••• •• Vaccine schedule
•• •
•••••••••••••••• •••••••
Neonatal
80 Infant

•••••••••••
60 •••
•••••
Vaccine efficacy

40

20

−20
0 50 100 150 0 50 100 150
Time since final dose of vaccination (weeks) Time since final dose of vaccination (weeks)

Figure 2: Median and 95% credible intervals of cumulative and instantaneous vaccine efficacy by duration of follow-up and type of schedule (neonatal vs
infant) following three doses of RV3-BB in Indonesia
A simple power function was used to represent vaccine waning over time; equivalent plots based on other potential waning functions are available in the appendix
(p 19). Data points shown on the left-hand panel represent observed vaccine efficacies derived from cumulative Kaplan-Meier hazard ratios, and error bars with their
corresponding 95% confidence intervals. Solid lines and dashed lines represent medians. Shaded areas represent 95% credible intervals.

A simple power function was fitted in all strata because schedules of Rotarix in South Africa and Malawi. We were
this required the fewest assumptions and parameters and unable to obtain the underlying dataset for this trial. In
had goodness of fit (DIC scores) that were consistently both countries, a three-dose schedule (administered at
favourable across all strata of interest compared with 6 weeks, 10 weeks, and 14 weeks) had higher VE than the
other functions (appendix p 8). Results for alternative two-dose schedule (administered at 10 weeks and
functions are shown in the appendix (p 10). 14 weeks), but the CIs were wide (table 1).
We found few RCTs with head-to-head comparisons of
different schedules. In Indonesia, a three-dose neonatal Discussion
RV3-BB schedule (administered at 0–5 days, 8–10 weeks, Our analysis showed that live oral rotavirus vaccines
and 14–16 weeks) was compared with a three-dose RV3-BB provide high and durable protection in low-mortality and
infant schedule (administered at 8–10 weeks, 14–16 weeks, medium-mortality settings. Efficacy is lower and wanes
and 18–20 weeks).15 For the neonatal schedule, the VE was more rapidly in high-mortality settings, but in these
94% (95% confidence interval 55–99) after about 9 months settings, more than 60% of rotavirus gastroenteritis
of follow-up and 75% (43–89) after about 15 months. For hospital admissions occur before age 1 year, and more
the infant schedule, VE was 77% (32–92) after about than 90% occur before age 2 years.51 Thus, live oral
8 months and 51% (7–74) after about 14 months (table 1). rotavirus vaccines are still likely to provide substantial
For this trial, we were able to obtain the number of events benefit in these settings, irrespective of waning.
in each week of follow-up to better inform estimates of iVE The reasons for lower rotavirus vaccine efficacy in
over time. For the neonatal schedule, the estimated iVE resource-poor settings are not well understood.
was 98% (92–100) after 2 weeks, 77% (73–80) after 6 months, Immunogenicity studies have shown lower geometric
and 57% (42–69) after 12 months of follow-up. For the mean concentrations in resource-poor settings than in
standard infant schedule, iVE was 95% (89–98) after high-income settings.52 Hypotheses for lower immuno-​
2 weeks, 60% (55–64) after 6 months, and 31% (12–48) after genicity include interference by maternal antibodies,
12 months of follow-up (figure 2). We found no significant interference by oral polio vaccines, neutralising factors
difference between the two schedules after 2 weeks of present in breastmilk, malnutrition, other enteric
follow-up, but strong evidence that the neonatal schedule coinfections, rotavirus strain diversity, and HIV infection.
had higher iVE after 6 months and 12 months of follow-up. Competition in the gut has also been proposed as a reason
The median difference in iVE was 3% (–1 to 10, p=0·088) for the lower performance of oral polio vaccine in
after 2 weeks of follow-up, 17% (13 to 22, p=0·00049) after resource-poor settings.53–55 Research is underway to assess
6 months, and 26% (13 to 48, p=0·017) after 12 months. the role of maternal antibodies and gut microbiota in the
We used a simple power function for the head-to-head immune response to rotavirus vaccines in British,
analyses because it required the minimum number of Malawian, and Indian infants.56 Two pivotal cohort studies
assumptions and parameters and had favourable DIC from Mexico57 and India58 have reported contrasting
scores (appendix p 8). Results for alternative functions estimates of the protection conferred by natural infections
are also shown in the appendix (p 19). against subsequent disease. In Mexico (a medium-
The only other trial with head-to-head comparison of mortality setting), two previous infections (asymptomatic
schedules was a multicountry trial comparing infant or symptomatic) conferred 100% protection against

www.thelancet.com/infection Vol 19 July 2019 723


Articles

subsequent moderate or severe rotavirus gastroenteritis. and Rotarix, but the Rotarix sites included data points
In India (a high-mortality setting), the equivalent pro-​ from South Africa, which had higher efficacy and lower
tection was 57% after two previous infections (and 79% child mortality relative to the sites evaluated in the
after three previous infections). If natural infections are RotaTeq trials. Thus, in the absence of head-to-head
less likely to protect against moderate and severe rotavirus comparisons from the same trial populations, there is
gastroenteritis in higher-mortality settings than in lower- insufficient evidence to favour one product over another
mortality settings, then a live oral vaccine mimicking in terms of vaccine efficacy and duration of protection.
natural infection might also have lower estimated efficacy However, the postlicensure experience of countries that
in children in these settings. have used both Rotarix and RotaTeq does not suggest any
The reported declines in instantaneous efficacy might material difference in vaccine effect.63
not be entirely caused by declining vaccine-induced Most of the data points were reported against the
antibodies. Some of the decrease could be explained by a Vesikari 11–20 scale, but some were reported against the
higher incidence of natural asymptomatic and mild Clark 16–24 scale. These two scores correlate poorly with
infections (and thus preferential immune boosting) one another when estimating the proportion of rotavirus
among unvaccinated controls compared with vaccine gastroenteritis episodes defined as severe.64,65 However,
recipients. In these circumstances the risk of severe this bias is unlikely to change the conclusion that
rotavirus gastroenteritis in vaccine recipients would protection is high and durable in the low-mortality
gradually converge with, and might exceed, the risk in stratum, in which the Clark scale was more commonly
unvaccinated controls over time. This phenomenon has used.
been described previously in the context of so-called We stratified our results by mortality and presented
leaky vaccines.59 Our analysis of the infant and neonatal pooled results with and without data points from
schedules for RV3-BB in Indonesia suggested a positive potentially influential studies. We restricted the analysis
protective effect of the vaccine in the first 18 months of to RCTs because they represent the gold standard
follow-up, but extrapolation of the curves suggested a approach for measuring per-protocol vaccine efficacy and
negative effect thereafter. This would be consistent provide accurate information about the mean duration of
with preferential natural boosting among non-vaccine follow-up. Other designs, such as case-control studies, do
recipients but is speculative, because it involves extra-​ not permit precise estimation of the mean duration of
polating beyond the observed period of follow-up in the follow-up. Some case-control studies report vaccine
trial. Reanalysis of data from an RCT in Bangladesh60 has effectiveness by age band, and thus could potentially be
allowed these effects to be partly disentangled by used to derive the duration of follow-up, but this
excluding any children who had an episode of non-severe approach becomes increasingly crude as the width of the
rotavirus gastroenteritis. This finding explained some, age band increases. Case-control studies are also at risk
but not all, of the reduction in vaccine efficacy over time. of bias because infants who have been vaccinated might
However, it was not possible to exclude infants who had be different from those who are unvaccinated for both
previous asymptomatic infections, which might also known and unknown reasons. We also restricted the
have an important role. analysis to per protocol rather than by intention to treat
Head-to-head comparisons of schedules for the same because this analysis provided a consistent basis for
vaccine were rare, and more evidence is needed from pooling the different RCTs, ensuring that all infants
more places on the relative benefits of one schedule over received the recommended number of doses and that a
another. In our analysis of RV3-BB in Indonesia, the more consistent starting point was used for the
neonatal schedule provided more durable protection measurement of follow-up. Accurate estimates of iVE
than the infant schedule, but this analysis was based on following a single dose of rotavirus vaccination would be
few case counts in each week of follow-up. A neonatal useful for informing the potential effects of different
schedule is also likely to result in higher and earlier schedules, but typically there are few infants who receive
coverage and fewer vaccine-related intussusception only a single dose, and even fewer of those infants are
events than an infant schedule, so warrants serious followed up for the full duration of the trial.
consideration. Other strategies that could help to improve We reported the initial peak efficacy starting at 2 weeks
the effect of the vaccines include administering a booster of follow-up because of uncertainty around the time that
dose later in infancy17,61 or using injectable non-replicating antibodies might take to develop after vaccination. In
vaccines,62 but more evidence is needed on the safety and addition, we had to extrapolate our fitted estimates of VE
clinical efficacy of both of these options. to periods without empirical data (eg, beyond 2 years of
For the pooled analysis, we combined evidence for follow-up). The absence of empirical data from RCTs is
different vaccine products and different infant schedules represented by larger credible intervals in these periods.
to avoid having small numbers of data points in each However, this makes comparison of different waning
stratum. None of the RCTs compared different brands functions difficult. Evidence from RCTs with a longer
of rotavirus vaccination head to head in the same duration of follow-up or high-quality observational
population. There were several observations for RotaTeq studies is needed to overcome this knowledge gap. A

724 www.thelancet.com/infection Vol 19 July 2019


Articles

so-called no-waning model required only one parameter, vaccines in different settings. Our analysis provides the
and for this reason had favourable DIC scores in each of most comprehensive evidence to date that rotavirus
the pooled analyses. However, in Indonesia, where all vaccine efficacy is lower and wanes more rapidly in high-
data points were from the same trial, the DIC score was mortality settings than in low-mortality settings. The
unfavourable. We considered no waning (or no change in earlier peak age of disease in these settings means that
RR) to be implausible, given that VE was shown to live oral rotavirus vaccines are still likely to provide
decrease in nearly all RCTs with more than one follow-up substantial benefit, but strategies with the potential to
point. However, more studies with multiple follow-up further increase the effects of vaccines, such as neonatal
times would be needed to have greater certainty about vaccination, warrant serious consideration. Monitoring
the appropriate form of vaccine waning. the age distribution of rotavirus disease cases in the years
We used a novel approach to convert estimates from following vaccine introduction will also be important.
cumulative VE to iVE. We show that this method is able Consistent with the basic theory of infectious disease
to retrieve iVE, and that standard estimates of dynamics, a reduction in the incidence of infection (eg,
cumulative VE might overestimate iVE in the presence from vaccination) should lead to an increase in the mean
of waning. However, there are some limitations in age of infection. As more children become infected at
applying this method to the meta-regression used in older ages, the need for more durable rotavirus vaccines
this study. First, it would be better to use relative rates might become more pressing.
than RR. We had to compute relative risks because most Contributors
of the RCTs only reported observed numbers of cases AC devised the idea for the study, extracted the data, ran preliminary
and individuals at a limited number of follow-up times. statistical analyses, and wrote the first draft of the paper. KVZ developed
the methods for converting cumulative vaccine efficacy into instantaneous
However, because severe rotavirus gastroenteritis is a efficacy, ran the Bayesian meta-regressions, and designed the figures.
relatively rare outcome, risk ratios and rate ratios are SF, CS and MJ ran preliminary statistical analyses and provided
expected to be similar, and we assume that this bias is conceptual and methodological guidance. JB, JT and UP contributed to
negligible in our study. Second, waning of vaccine data interpretation. All authors provided comments on the draft.
efficacy (or conversely, waxing of the relative rate) might Declaration of interests
interact with changes in baseline rates. This effect JB reports project grants from the Bill & Melinda Gates Foundation and
the National Health and Medical Research Council for the conduct of
would be relatively small on the estimated VE, but clinical trials of the RV3-BB rotavirus vaccine at MCRI, and other
might be pronounced when converted to an iVE. support from the Victorian Government Operational Infrastructure
Because we had no information on changes in the Support Program. All other authors declare no competing interests.
baseline rates in our studies, we assumed that this rate Acknowledgments
was constant over time (an assumption that is often This work was supported by the Bill & Melinda Gates Foundation
made in survival analyses) and did not correct for it. The (grant number OPP1147721) and the Vaccine Impact Modelling
Consortium (grant number OPP1157270). The views expressed are those
bias is likely to be in the direction of increasing VE of the authors and not necessarily those of the Consortium or its
because baseline rates decline with age, particularly in funders. SF is funded through a Sir Henry Dale Fellowship jointly
high-mortality settings. Our method should ideally be funded by the Wellcome Trust and the Royal Society (grant number
208812/Z/17/Z). We would like to acknowledge the WHO Immunization
extended to control for different changing baseline rates
and Vaccines Related Implementation Research Advisory Committee for
in different studies, as would be the case in a pooled providing useful feedback on the analysis. We also thank Cochrane
analysis. However, even uncorrected iVE should still be Response (Hanna Bergman, Nicholas Henschke, and
a better approximation to true iVE than cumulative VE Karla Soares-Weiser) for sharing the list of RCTs identified in their
updated systematic review. The findings and conclusions in this report
in the presence of waning.
are those of the authors and do not necessarily represent the official
Because the two Indian studies had larger sample sizes position of the Centers for Disease Control and Prevention.
than other studies in the high-mortality stratum, we
References
did a sensitivity analysis to see whether these studies 1 GBD Diarrhoeal Diseases Collaborators. Estimates of global, regional,
significantly influenced our results. Exclusion of these and national morbidity, mortality, and aetiologies of diarrhoeal
diseases: a systematic analysis for the Global Burden of Disease Study
studies did not alter our findings significantly. Moreover, 2015. Lancet Infect Dis 2017; 17: 909–48.
an analysis in which we estimated iVE for India alone did 2 Lanata CF, Fischer-Walker CL, Olascoaga AC, et al. Global causes of
not provide meaningful results, because there were only diarrheal disease mortality in children <5 years of age: a systematic
review. PLoS One 2013; 8: e72788.
four observations, which is too few to generate a reliable
3 Tate JE, Burton AH, Boschi-Pinto C, Parashar UD, World Health
pooled estimate in our analysis. Organization-Coordinated Global Rotavirus Surveillance Network.
Reviews of the efficacy, effectiveness, and effects of Global, regional, and national estimates of rotavirus mortality in
children <5 years of age, 2000–2013. Clin Infect Dis 2016;
rotavirus vaccines19,66,67 have described variation in rotavirus 62 (suppl 2): S96–105.
vaccine effects according to under-5 mortality and 4 Rheingans RD, Antil L, Dreibelbis R, Podewils LJ, Bresee JS,
geographical region. However, to our knowledge, our Parashar UD. Economic costs of rotavirus gastroenteritis and
study is the first to synthesise all the available RCT evidence cost-effectiveness of vaccination in developing countries. J Infect Dis
2009; 200 (suppl 1): S16–27.
and to obtain robust estimates of iVE by duration of 5 Thomas SL, Walker JL, Fenty J, et al. Impact of the national
follow-up. Our study should provide important evidence rotavirus vaccination programme on acute gastroenteritis in
for estimating and monitoring the effects of rotavirus England and associated costs averted. Vaccine 2017; 35: 680–86.

www.thelancet.com/infection Vol 19 July 2019 725


Articles

6 Rota Council. Country introductions of rotavirus vaccines. 28 GitHub. R code. https://github.com/kevinvzandvoort/rotavirus_


http://rotacouncil.org/vaccine-introduction/global-introduction- vaccine_efficacy (accessed Jan 15, 2019).
status/ (accessed Aug 31, 2018). 29 Armah GE, Sow SO, Breiman RF, et al. Efficacy of pentavalent
7 Ruiz-Palacios GM, Perez-Schael I, Velazquez FR, et al. Safety and rotavirus vaccine against severe rotavirus gastroenteritis in infants in
efficacy of an attenuated vaccine against severe rotavirus developing countries in sub-Saharan Africa: a randomised,
gastroenteritis. N Engl J Med 2006; 354: 11–22. double-blind, placebo-controlled trial. Lancet 2010; 376: 606–14.
8 Vesikari T, Matson DO, Dennehy P, et al. Safety and efficacy of a 30 Li RC, Huang T, Li Y, et al. Human rotavirus vaccine (RIX4414)
pentavalent human-bovine (WC3) reassortant rotavirus vaccine. efficacy in the first two years of life: a randomized, placebo-controlled
N Engl J Med 2006; 354: 23–33. trial in China. Hum Vaccin Immunother 2014; 10: 11–18.
9 Colgate ER, Haque R, Dickson DM, et al. Delayed dosing of oral 31 Salinas B, Perez Schael I, Linhares AC, et al. Evaluation of safety,
rotavirus vaccine demonstrates decreased risk of rotavirus immunogenicity and efficacy of an attenuated rotavirus vaccine,
gastroenteritis associated with serum zinc: a randomized controlled RIX4414: a randomized, placebo-controlled trial in Latin American
trial. Clin Infect Dis 2016; 63: 634–41. infants. Pediatr Infect Dis J 2005; 24: 807–16.
10 Cunliffe NA, Witte D, Ngwira BM, et al. Efficacy of human rotavirus 32 Tregnaghi MW, Abate HJ, Valencia A, et al. Human rotavirus
vaccine against severe gastroenteritis in Malawian children in the vaccine is highly efficacious when coadministered with routine
first two years of life: a randomized, double-blind, placebo expanded program of immunization vaccines including oral
controlled trial. Vaccine 2012; 30 (suppl 1): A36–43. poliovirus vaccine in Latin America. Pediatr Infect Dis J 2011;
11 Madhi SA, Kirsten M, Louw C, et al. Efficacy and immunogenicity 30: e103–08.
of two or three dose rotavirus-vaccine regimen in South African 33 Linhares AC, Velazquez FR, Perez-Schael I, et al. Efficacy and safety
children over two consecutive rotavirus-seasons: a randomized, of an oral live attenuated human rotavirus vaccine against rotavirus
double-blind, placebo-controlled trial. Vaccine 2012; gastroenteritis during the first 2 years of life in Latin American
30 (suppl 1): A44–51. infants: a randomised, double-blind, placebo-controlled phase III
12 Zaman K, Dang DA, Victor JC, et al. Efficacy of pentavalent study. Lancet 2008; 371: 1181–89.
rotavirus vaccine against severe rotavirus gastroenteritis in infants 34 Mo Z, Mo Y, Li M, et al. Efficacy and safety of a pentavalent live
in developing countries in Asia: a randomised, double-blind, human-bovine reassortant rotavirus vaccine (RV5) in healthy
placebo-controlled trial. Lancet 2010; 376: 615–23. Chinese infants: a randomized, double-blind, placebo-controlled trial.
13 Kulkarni PS, Desai S, Tewari T, et al. A randomized phase III Vaccine 2017; 35: 5897–04.
clinical trial to assess the efficacy of a bovine-human reassortant 35 Vesikari T, Itzler R, Matson DO, et al. Efficacy of a pentavalent
pentavalent rotavirus vaccine in Indian infants. Vaccine 2017; rotavirus vaccine in reducing rotavirus-associated health care
35: 6228–37. utilization across three regions (11 countries). Int J Infect Dis 2007;
14 Isanaka S, Guindo O, Langendorf C, et al. Efficacy of a low-cost, 11 (suppl 2): S29–35.
heat-stable oral rotavirus vaccine in Niger. N Engl J Med 2017; 36 Grant LR, Watt JP, Weatherholtz RC, et al. Efficacy of a pentavalent
376: 1121–30. human-bovine reassortant rotavirus vaccine against rotavirus
15 Bines JE, At Thobari J, Satria CD, et al. Human neonatal rotavirus gastroenteritis among American Indian children. Pediatr Infect Dis J
vaccine (RV3-BB) to target rotavirus from birth. N Engl J Med 2018; 2012; 31: 184–88.
378: 719–30. 37 Vesikari T, Karvonen A, Prymula R, et al. Efficacy of human rotavirus
16 Bhandari N, Rongsen-Chandola T, Bavdekar A, et al. Efficacy of vaccine against rotavirus gastroenteritis during the first 2 years of life
a monovalent human-bovine (116E) rotavirus vaccine in Indian in European infants: randomised, double-blind controlled study.
infants: a randomised, double-blind, placebo-controlled trial. Lancet 2007; 370: 1757–63.
Lancet 2014; 383: 2136–43. 38 Vesikari T, Karvonen A, Puustinen L, et al. Efficacy of RIX4414 live
17 Haidara FC, Tapia MD, Sow SO, et al. Evaluation of a booster dose attenuated human rotavirus vaccine in Finnish infants.
of pentavalent rotavirus vaccine coadministered with measles, Pediatr Infect Dis J 2004; 23: 937–43.
yellow fever, and meningitis a vaccines in 9-month-old Malian 39 Kawamura N, Tokoeda Y, Oshima M, et al. Efficacy, safety and
infants. J Infect Dis 2018; 218: 606–13. immunogenicity of RIX4414 in Japanese infants during the first two
18 Soares-Weiser K, Bergman H, Henschke N, Pitan F, Cunliffe N. years of life. Vaccine 2011; 29: 6335–41.
Vaccines for preventing rotavirus diarrhoea: vaccines in use. 40 Phua KB, Lim FS, Lau YL, et al. Rotavirus vaccine RIX4414 efficacy
Coch Data Syst Rev 2019; published online March 25. sustained during the third year of life: a randomized clinical trial in
DOI:10.1002/14651858.CD008521.pub4. an Asian population. Vaccine 2012; 30: 4552–57.
19 Lamberti LM, Ashraf S, Walker CL, Black RE. A systematic review 41 Bernstein DI, Sack DA, Rothstein E, et al. Efficacy of live, attenuated,
of the effect of rotavirus vaccination on diarrhea outcomes among human rotavirus vaccine 89–12 in infants: a randomised
children younger than 5 Years. Pediatr Infect Dis J 2016; 35: 992–98. placebo-controlled trial. Lancet 1999; 354: 287–90.
20 Ruuska T, Vesikari T. Rotavirus disease in Finnish children: use of 42 Vesikari T, Itzler R, Karvonen A, et al. RotaTeq, a pentavalent
numerical scores for clinical severity of diarrhoeal episodes. rotavirus vaccine: efficacy and safety among infants in Europe.
Scand J Infect Dis 1990; 22: 259–67. Vaccine 2010; 28: 345–51.
21 Clark HF, Borian FE, Bell LM, Modesto K, Gouvea V, Plotkin SA. 43 Block SL, Vesikari T, Goveia MG, et al. Efficacy, immunogenicity, and
Protective effect of WC3 vaccine against rotavirus diarrhea in safety of a pentavalent human-bovine (WC3) reassortant rotavirus
infants during a predominantly serotype 1 rotavirus season. vaccine at the end of shelf life. Pediatrics 2007; 119: 11–18.
J Infect Dis 1988; 158: 570–87. 44 Iwata S, Nakata S, Ukae S, et al. Efficacy and safety of
22 UN, Department of Economic and Social Affairs, Population pentavalent rotavirus vaccine in Japan: a randomized, double-blind,
Division (2017). world population prospects: the 2017 revision, placebo-controlled, multicenter trial. Hum Vaccin Immunother 2013;
DVD edition. http://esa.un.org/wpp/ (accessed Oct 1, 2018). 9: 1626–33.
23 Altman DG. Confidence intervals for the number needed to treat. 45 Clark HF, Bernstein DI, Dennehy PH, et al. Safety, efficacy, and
BMJ 1998; 317: 1309–12. immunogenicity of a live, quadrivalent human-bovine reassortant
24 Daly LE. Confidence limits made easy: interval estimation using rotavirus vaccine in healthy infants. J Pediatr 2004; 144: 184–90.
a substitution method. Am J Epidemiol 1998; 147: 783–90. 46 Feikin DR, Laserson KF, Ojwando J, et al. Efficacy of pentavalent
25 Deeks JJ, Higgins JPT. Statistical algorithms in review manager 5.1. rotavirus vaccine in a high HIV prevalence population in Kenya.
https://training.cochrane.org/handbook/statistical-methods- Vaccine 2012; 30 (suppl 1): A52–60.
revman5 (accessed Sept 1, 2018). 47 Vesikari T, Prymula R, Schuster V, et al. Efficacy and immunogenicity
26 R: a language and environment for statistical computing. of live-attenuated human rotavirus vaccine in breast-fed and
R Foundation for Statistical Computing, Vienna, Austria. URL formula-fed European infants. Pediatr Infect Dis J 2012; 31: 509–13.
http://www.R-project.org/. 48 Vesikari T, Karvonen A, Ferrante SA, Ciarlet M. Efficacy of the
27 Plummer M. rjags: Bayesian graphical models using MCMC. pentavalent rotavirus vaccine, RotaTeq(R), in Finnish infants up to
R package version 4-6. https://CRAN.R-project.org/package=rjags 3 years of age: the Finnish Extension Study. Eur J Pediatr 2010;
(accessed March 1, 2019). 169: 1379–86.

726 www.thelancet.com/infection Vol 19 July 2019


Articles

49 Vesikari T, Karvonen A, Ferrante SA, Kuter BJ, Ciarlet M. 59 Smith PG, Rodrigues LC, Fine PE. Assessment of the protective
Sustained efficacy of the pentavalent rotavirus vaccine, RV5, up to efficacy of vaccines against common diseases using case-control
3.1 years following the last dose of vaccine. Pediatr Infect Dis J 2010; and cohort studies. Int J Epidemiol 1984; 13: 87–93.
29: 957–63. 60 Rogawski ET, Platts-Mills JA, Colgate ER, et al. Quantifying the
50 Armah GE, Kapikian AZ, Vesikari T, et al. Efficacy, immunogenicity, impact of natural immunity on rotavirus vaccine efficacy estimates:
and safety of two doses of a tetravalent rotavirus vaccine RRV-TV in a clinical trial in Dhaka, Bangladesh (PROVIDE) and a simulation
Ghana with the first dose administered during the neonatal period. study. J Infect Dis 2018; 217: 861–68.
J Infect Dis 2013; 208: 423–31. 61 Burnett E, Lopman BA, Parashar UD. Potential for a booster dose of
51 Hasso-Agopsowicz M, Nanji Ladva C, Lopman B, et al. Global rotavirus vaccine to further reduce diarrhea mortality. Vaccine 2017;
review of the age distribution of rotavirus disease in children aged 35: 7198–03.
<5 years before the introduction of rotavirus vaccination. 62 Kirkwood CD, Ma LF, Carey ME, Steele AD. The rotavirus vaccine
Clin Infect Dis 2019; published Jan 28. DOI:10.1093/cid/ciz060. development pipeline. Vaccine 2017; published online April 7.
52 Patel M, Glass RI, Jiang B, Santosham M, Lopman B, Parashar U. DOI:10.1016/j.vaccine.2017.03.076.
A systematic review of anti-rotavirus serum IgA antibody titer as 63 Immergluck LC, Parker TC, Jain S, et al. Sustained effectiveness of
a potential correlate of rotavirus vaccine efficacy. J Infect Dis 2013; monovalent and pentavalent rotavirus vaccines in children. J Pediatr
208: 284–94. 2016; 172: 116–20.
53 Chan J, Nirwati H, Triasih R, et al. Maternal antibodies to rotavirus: 64 Aslan A, Kurugol Z, Cetin H, Karakaslilar S, Koturoglu G.
could they interfere with live rotavirus vaccines in developing Comparison of Vesikari and Clark scales regarding the definition of
countries? Vaccine 2011; 29: 1242–47. severe rotavirus gastroenteritis in children. Infect Dis 2015;
54 Lopman BA, Pitzer VE. Waxing understanding of waning 47: 332–37.
immunity. J Infect Dis 2018; 217: 851–53. 65 Givon-Lavi N, Greenberg D, Dagan R. Comparison between
55 Taniuchi M, Platts-Mills JA, Begum S, et al. Impact of enterovirus two severity scoring scales commonly used in the evaluation of
and other enteric pathogens on oral polio and rotavirus vaccine rotavirus gastroenteritis in children. Vaccine 2008; 26: 5798–801.
performance in Bangladeshi infants. Vaccine 2016; 34: 3068–75. 66 Jonesteller CL, Burnett E, Yen C, Tate JE, Parashar UD.
56 Sindhu KN, Cunliffe N, Peak M, et al. Impact of maternal Effectiveness of rotavirus vaccination: a systematic review of the
antibodies and infant gut microbiota on the immunogenicity of first decade of global post-licensure data, 2006–2016. Clin Infect Dis
rotavirus vaccines in African, Indian and European infants: protocol 2017; 65: 840–50.
for a prospective cohort study. BMJ Open 2017; 7: e016577. 67 Patel MM, Clark AD, Sanderson CF, Tate J, Parashar UD.
57 Velazquez FR, Matson DO, Calva JJ, et al. Rotavirus infection in Removing the age restrictions for rotavirus vaccination:
infants as protection against subsequent infections. N Engl J Med a benefit-risk modeling analysis. PLoS Med 2012; 9: e1001330.
1996; 335: 1022–28.
58 Gladstone BP, Ramani S, Mukhopadhya I, et al. Protective effect of
natural rotavirus infection in an Indian birth cohort. N Engl J Med
2011; 365: 337–46.

www.thelancet.com/infection Vol 19 July 2019 727

You might also like